Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Gastroenterology ; 156(6): 1788-1804.e13, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30641053

RESUMEN

BACKGROUND & AIMS: Patients with cirrhosis are at high risk for hepatocellular carcinoma (HCC) and often have increased serum levels of estrogen. It is not clear how estrogen promotes hepatic growth. We investigated the effects of estrogen on hepatocyte proliferation during zebrafish development, liver regeneration, and carcinogenesis. We also studied human hepatocytes and liver tissues. METHODS: Zebrafish were exposed to selective modifiers of estrogen signaling at larval and adult stages. Liver growth was assessed by gene expression, fluorescent imaging, and histologic analyses. We monitored liver regeneration after hepatocyte ablation and HCC development after administration of chemical carcinogens (dimethylbenzanthrazene). Proliferation of human hepatocytes was measured in a coculture system. We measured levels of G-protein-coupled estrogen receptor (GPER1) in HCC and nontumor liver tissues from 68 patients by immunohistochemistry. RESULTS: Exposure to 17ß-estradiol (E2) increased proliferation of hepatocytes and liver volume and mass in larval and adult zebrafish. Chemical genetic and epistasis experiments showed that GPER1 mediates the effects of E2 via the phosphoinositide 3-kinase-protein kinase B-mechanistic target of rapamycin pathway: gper1-knockout and mtor-knockout zebrafish did not increase liver growth in response to E2. HCC samples from patients had increased levels of GPER1 compared with nontumor tissue samples; estrogen promoted proliferation of human primary hepatocytes. Estrogen accelerated hepatocarcinogenesis specifically in male zebrafish. Chemical inhibition or genetic loss of GPER1 significantly reduced tumor development in the zebrafish. CONCLUSIONS: In an analysis of zebrafish and human liver cells and tissues, we found GPER1 to be a hepatic estrogen sensor that regulates liver growth during development, regeneration, and tumorigenesis. Inhibitors of GPER1 might be developed for liver cancer prevention or treatment. TRANSCRIPT PROFILING: The accession number in the Gene Expression Omnibus is GSE92544.


Asunto(s)
Carcinoma Hepatocelular/metabolismo , Estradiol/farmacología , Estrógenos/farmacología , Neoplasias Hepáticas/metabolismo , Hígado/crecimiento & desarrollo , Receptores de Estrógenos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Proteínas de Pez Cebra/metabolismo , 9,10-Dimetil-1,2-benzantraceno , Animales , Carcinogénesis/efectos de los fármacos , Carcinoma Hepatocelular/patología , Proliferación Celular/efectos de los fármacos , Femenino , Expresión Génica/efectos de los fármacos , Hepatocitos , Humanos , Hígado/metabolismo , Cirrosis Hepática/metabolismo , Neoplasias Hepáticas/patología , Regeneración Hepática , Masculino , Tamaño de los Órganos/efectos de los fármacos , Fosfatidilinositol 3-Quinasa/metabolismo , Receptores Acoplados a Proteínas G/genética , Factores Sexuales , Transducción de Señal , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo , Carga Tumoral/efectos de los fármacos , Pez Cebra , Proteínas de Pez Cebra/genética
2.
EMBO J ; 31(14): 3079-91, 2012 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-22692129

RESUMEN

Two types of stem cells are currently defined in small intestinal crypts: cycling crypt base columnar (CBC) cells and quiescent '+4' cells. Here, we combine transcriptomics with proteomics to define a definitive molecular signature for Lgr5(+) CBC cells. Transcriptional profiling of FACS-sorted Lgr5(+) stem cells and their daughters using two microarray platforms revealed an mRNA stem cell signature of 384 unique genes. Quantitative mass spectrometry on the same cell populations identified 278 proteins enriched in intestinal stem cells. The mRNA and protein data sets showed a high level of correlation and a combined signature of 510 stem cell-enriched genes was defined. Spatial expression patterns were further characterized by mRNA in-situ hybridization, revealing that approximately half of the genes were expressed in a gradient with highest levels at the crypt bottom, while the other half was expressed uniquely in Lgr5(+)stem cells. Lineage tracing using a newly established knock-in mouse for one of the signature genes, Smoc2, confirmed its stem cell specificity. Using this resource, we find-and confirm by independent approaches-that the proposed quiescent/'+4' stem cell markers Bmi1, Tert, Hopx and Lrig1 are robustly expressed in CBC cells.


Asunto(s)
Antígenos de Diferenciación/metabolismo , Regulación de la Expresión Génica , Mucosa Intestinal/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Células Madre/metabolismo , Animales , Antígenos de Diferenciación/genética , Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/metabolismo , Perfilación de la Expresión Génica , Intestinos/citología , Ratones , Ratones Transgénicos , Análisis de Secuencia por Matrices de Oligonucleótidos , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Receptores Acoplados a Proteínas G/genética , Células Madre/citología
3.
EMBO Rep ; 15(1): 62-9, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24355609

RESUMEN

The concept of 'field cancerization' describes the clonal expansion of genetically altered, but morphologically normal cells that predisposes a tissue to cancer development. Here, we demonstrate that biased stem cell competition in the mouse small intestine can initiate the expansion of such clones. We quantitatively analyze how the activation of oncogenic K-ras in individual Lgr5(+) stem cells accelerates their cell division rate and creates a biased drift towards crypt clonality. K-ras mutant crypts then clonally expand within the epithelium through enhanced crypt fission, which distributes the existing Paneth cell niche over the two new crypts. Thus, an unequal competition between wild-type and mutant intestinal stem cells initiates a biased drift that leads to the clonal expansion of crypts carrying oncogenic mutations.


Asunto(s)
Células Madre Adultas/fisiología , Neoplasias Colorrectales/patología , Proteínas Proto-Oncogénicas p21(ras)/genética , Receptores Acoplados a Proteínas G/metabolismo , Animales , Ciclo Celular , Proliferación Celular , Transformación Celular Neoplásica , Neoplasias Colorrectales/genética , Mucosa Intestinal/patología , Intestino Delgado/patología , Ratones , Ratones Transgénicos , Mutación Missense , Oncogenes , Receptores Acoplados a Proteínas G/genética , Nicho de Células Madre
4.
Angew Chem Int Ed Engl ; 55(40): 12440-4, 2016 09 26.
Artículo en Inglés | MEDLINE | ID: mdl-27554600

RESUMEN

The ability to remotely trigger CRISPR/Cas9 activity would enable new strategies to study cellular events with greater precision and complexity. In this work, we have developed a method to photocage the activity of the guide RNA called "CRISPR-plus" (CRISPR-precise light-mediated unveiling of sgRNAs). The photoactivation capability of our CRISPR-plus method is compatible with the simultaneous targeting of multiple DNA sequences and supports numerous modifications that can enable guide RNA labeling for use in imaging and mechanistic investigations.


Asunto(s)
Sistemas CRISPR-Cas/genética , ARN Guía de Kinetoplastida/metabolismo , Secuencia de Bases , Proteínas Fluorescentes Verdes/genética , Células HeLa , Humanos , Luz , Hibridación de Ácido Nucleico , Fotólisis/efectos de la radiación , ARN Guía de Kinetoplastida/química
5.
EMBO J ; 30(6): 1104-9, 2011 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-21297579

RESUMEN

Somatic cells have been proposed to be limited in the number of cell divisions they can undergo. This is thought to be a mechanism by which stem cells retain their integrity preventing disease. However, we have recently discovered intestinal crypt stem cells that persist for the lifetime of a mouse, yet divide every day. We now demonstrate biochemically that primary isolated Lgr5+ve stem cells contain significant telomerase activity. Telomerase activity rapidly decreases in the undifferentiated progeny of these stem cells and is entirely lost in differentiated villus cells. Conversely, asymmetric segregation of chromosomes has been proposed as a mechanism for stem cells to protect their genomes against damage. We determined the average cell cycle length of Lgr5+ve stem cells at 21.5 h and find that Lgr5+ve intestinal stem cells randomly segregate newly synthesized DNA strands, opposing the 'immortal strand' hypothesis.


Asunto(s)
Segregación Cromosómica , Mucosa Intestinal/citología , Mucosa Intestinal/enzimología , Nicho de Células Madre , Células Madre/enzimología , Células Madre/fisiología , Telomerasa/metabolismo , Animales , Ciclo Celular , Diferenciación Celular , Células Epiteliales/enzimología , Células Epiteliales/fisiología , Ratones , Factores de Tiempo
6.
Carcinogenesis ; 35(1): 237-46, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23996931

RESUMEN

Although Apc mutation is widely considered an initiating event in colorectal cancer, little is known about the earliest stages of tumorigenesis following sporadic Apc loss. Therefore, we have utilized a novel mouse model that facilitates the sporadic inactivation of Apc via frameshift reversion of Cre in single, isolated cells and subsequently tracks the fates of Apc-deficient intestinal cells. Our results suggest that consistent with Apc being a 'gatekeeper', loss of Apc early in life during intestinal growth leads to adenomas or increased crypt fission, manifested by fields of mutant but otherwise normal-appearing crypts. In contrast, Apc loss occurring later in life has minimal consequences, with mutant crypts being less prone to either increased crypt fission or adenoma formation. Using the stem cell-specific Lgr5-CreER mouse, we generated different sized fields of Apc-deficient crypts via independent recombination events and found that field size correlates with progression to adenoma. To evaluate this early stage prior to adenoma formation as a therapeutic target, we examined the chemopreventive effects of sulindac on Apc-deficient occult crypt fission. We found that sulindac treatment started early in life inhibits the morphologically occult spread of Apc-deficient crypts and thus reduces adenoma numbers. Taken together these results suggest that: (i) earlier Apc loss promotes increased crypt fission, (ii) a field of Apc-deficient crypts, which can form via occult crypt fission or independent neighboring events, is an important intermediate between loss of Apc and adenoma formation and (iii) normal-appearing Apc-deficient crypts are potential unappreciated targets for cancer screening and chemoprevention.


Asunto(s)
Focos de Criptas Aberrantes/prevención & control , Adenoma/genética , Genes APC , Neoplasias Intestinales/genética , Sulindac/farmacología , Focos de Criptas Aberrantes/tratamiento farmacológico , Adenoma/patología , Factores de Edad , Animales , Quimioprevención , Reparación del ADN/genética , Neoplasias Intestinales/patología , Intestinos/patología , Ratones , Ratones Transgénicos , Mutación , Células Madre/patología , beta-Galactosidasa/genética , beta-Galactosidasa/metabolismo
7.
ACS Chem Biol ; 16(9): 1770-1778, 2021 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-34427427

RESUMEN

The utility of in vitro human disease models is mainly dependent on the availability and functional maturity of tissue-specific cell types. We have previously screened for and identified small molecules that can enhance hepatocyte function in vitro. Here, we characterize the functional effects of one of the hits, FH1, on primary human hepatocytes in vitro, and also in vivo on primary hepatocytes in a zebrafish model. Furthermore, we conducted an analogue screen to establish the structure-activity relationship of FH1. We performed affinity-purification proteomics that identified NQO2 to be a potential binding target for this small molecule, revealing a possible link between inflammatory signaling and hepatocellular function in zebrafish and human hepatocyte model systems.


Asunto(s)
Biomarcadores/metabolismo , Inhibidores Enzimáticos/química , Hepatocitos/metabolismo , Quinona Reductasas/antagonistas & inhibidores , Animales , Inhibidores Enzimáticos/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Ensayos Analíticos de Alto Rendimiento , Humanos , Interleucina-6/genética , Hígado , Simulación del Acoplamiento Molecular , Unión Proteica , Factor de Transcripción STAT3/genética , Transducción de Señal , Relación Estructura-Actividad , Factores de Necrosis Tumoral/genética , Pez Cebra
8.
Science ; 337(6095): 730-5, 2012 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-22855427

RESUMEN

The concept that tumors are maintained by dedicated stem cells, the so-called cancer stem cell hypothesis, has attracted great interest but remains controversial. Studying mouse models, we provide direct, functional evidence for the presence of stem cell activity within primary intestinal adenomas, a precursor to intestinal cancer. By "lineage retracing" using the multicolor Cre-reporter R26R-Confetti, we demonstrate that the crypt stem cell marker Lgr5 (leucine-rich repeat-containing heterotrimeric guanine nucleotide-binding protein-coupled receptor 5) also marks a subpopulation of adenoma cells that fuel the growth of established intestinal adenomas. These Lgr5(+) cells, which represent about 5 to 10% of the cells in the adenomas, generate additional Lgr5(+) cells as well as all other adenoma cell types. The Lgr5(+) cells are intermingled with Paneth cells near the adenoma base, a pattern reminiscent of the architecture of the normal crypt niche.


Asunto(s)
Adenoma/patología , Neoplasias Intestinales/patología , Células Madre Neoplásicas/patología , Células Madre Neoplásicas/fisiología , Receptores Acoplados a Proteínas G/análisis , Adenoma/genética , Adenoma/metabolismo , Animales , Biomarcadores/análisis , Linaje de la Célula , Transformación Celular Neoplásica , Perfilación de la Expresión Génica , Técnicas de Sustitución del Gen , Genes Reporteros , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Neoplasias Intestinales/genética , Ratones , Células Madre Multipotentes/patología , Células Madre Multipotentes/fisiología , Células de Paneth/patología , Nicho de Células Madre , Tamoxifeno/farmacología , Ensayo de Tumor de Célula Madre
9.
Nat Protoc ; 6(8): 1221-8, 2011 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-21799490

RESUMEN

In recent years, many mouse models have been developed to mark and trace the fate of adult cell populations using fluorescent proteins. High-resolution visualization of such fluorescent markers in their physiological setting is thus an important aspect of adult stem cell research. Here we describe a protocol to produce sections (150-200 µm) of near-native tissue with optimal tissue and cellular morphology by avoiding artifacts inherent in standard freezing or embedding procedures. The activity of genetically expressed fluorescent proteins is maintained, thereby enabling high-resolution three-dimensional (3D) reconstructions of fluorescent structures in virtually all types of tissues. The procedure allows immunofluorescence labeling of proteins to depths up to 50 µm, as well as a chemical 'Click-iT' reaction to detect DNA-intercalating analogs such as ethynyl deoxyuridine (EdU). Generation of near-native sections ready for imaging analysis takes approximately 2-3 h. Postsectioning processes, such as antibody labeling or EdU detection, take up to 10 h.


Asunto(s)
Proteínas Luminiscentes/análisis , Análisis de la Célula Individual/métodos , Procesamiento de Imagen Asistido por Computador , Imagenología Tridimensional , Microscopía Fluorescente/métodos , Microtomía
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA